Language selection

Search

Patent 2581814 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2581814
(54) English Title: STABLE TABLET FORMULATION OF TETRAHYDROBIOPTERIN
(54) French Title: FORMULE STABLE DE COMPRIMES
Status: Granted
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 9/20 (2006.01)
  • A61K 31/505 (2006.01)
  • A61P 3/00 (2006.01)
(72) Inventors :
  • JUNGLES, STEVEN (United States of America)
  • HENDERSON, MARK (United States of America)
  • SLUZKY, VICTORIA (United States of America)
  • BAFFI, ROBERT (United States of America)
(73) Owners :
  • BIOMARIN PHARMACEUTICAL INC. (United States of America)
(71) Applicants :
  • BIOMARIN PHARMACEUTICAL INC. (United States of America)
(74) Agent: KIRBY EADES GALE BAKER
(74) Associate agent:
(45) Issued: 2015-10-13
(86) PCT Filing Date: 2005-11-16
(87) Open to Public Inspection: 2006-05-26
Examination requested: 2010-11-10
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2005/041252
(87) International Publication Number: WO2006/055511
(85) National Entry: 2007-03-22

(30) Application Priority Data:
Application No. Country/Territory Date
60/629,189 United States of America 2004-11-17

Abstracts

English Abstract




The present invention is directed to a stable solid formulations of
tetrahydrobiopterin, processes for producing them, and treatment methods using
such formulations.


French Abstract

L'invention porte: sur des formules stables solides de tétrahydrobioptérine, sur leurs procédés de production, et sur des méthodes de traitement les utilisant. .

Claims

Note: Claims are shown in the official language in which they were submitted.


- 36 -
CLAIMS:
1. A stable tablet formulation comprising an initial amount of a
crystalline
polymorph, designated polymorph B, of (6R)-L-erythro-tetrahydrobiopterin, an
antioxidant, and a pharmaceutically acceptable excipient,
wherein the weight ratio of the antioxidant to (6R)-L-erythro-
tetrahydrobiopterin is in the range of about 1:5 to about 1:30;
wherein, after six months at room temperature and about 60% humidity,
the stable tablet formulation retains at least about 95% of the initial amount
of (6R)-L-
erythro-tetrahydrobiopterin; and
wherein said crystalline polymorph, as a hydrochloride salt, exhibits an X-
ray powder diffraction pattern with the following characteristic peaks
expressed in d-
values(.ANG.): 8.7 (vs), 5.63 (m), 4.76(m), 4.40 (m), 4.00 (s), 3.23 (s), and
3.11 (vs).
2. The stable tablet formulation of claim 1, wherein the crystalline
polymorph, as a hydrochloride salt, exhibits an X-ray powder diffraction
pattern with the
following characteristic peaks expressed in d-values(.ANG.): 8.7 (vs), 6.9
(w), 5.90 (vw), 5.63
(m), 5.07 (m), 4.76 (m), 4.40 (m), 4.15 (w), 4.00 (s), 3.95 (m), 3.52 (m),
3.44 (w), 3.32
(m), 3.23 (s), 3.17 (w), 3.11 (vs), 3.06 (w), 2.99 (w), 2.96 (w), 2.94 (m),
2.87 (w), 2.84
(s), 2.82 (m), 2.69 (w), 2.59 (w), and 2.44 (w).
3. The stable tablet formulation of claim 1 or 2, wherein, after nine
months at
room temperature and about 60% humidity, the stable tablet formulation retains
at least
about 95% of the initial amount of (6R)-L-erythro-tetrahydrobiopterin.
4. The stable tablet formulation of any one of claims 1 to 3, wherein the
stable tablet formulation retains at least about 98% of the initial amount of
(6R)-L-
erythro-tetrahydrobiopterin.
5. The stable tablet formulation of any one of claims 1 to 4, wherein the
initial amount of (6R)-L-erythro-tetrahydrobiopterin is in the range of about
30 wt% to
about 40 wt% of the formulation.

- 37 -
6. The stable tablet formulation of claim 5, wherein the initial amount of
(6R)-L-erythro-tetrahydrobiopterin is in the range of about 32 wt% to about 35
wt% of
the formulation.
7. The stable tablet formulation of claim 5, wherein the initial amount of
(6R)-L-erythro-tetrahydrobiopterin is about 33 wt%.
8. The stable tablet formulation of any one of claims 1 to 7, wherein the
initial amount of (6R)-L-erythro-tetrahydrobiopterin in a tablet is about 100
mg.
9. The stable tablet formulation of any one of claims 1 to 7, wherein the
initial amount of (6R)-L-erythro-tetrahydrobiopterin in a tablet is about 200
mg, about
300 mg, or about 400 mg.
10. The stable tablet formulation of any one of claims 1 to 9, further
comprising a binder.
11. The stable tablet formulation of claim 10, wherein the binder is
anhydrous
dibasic calcium phosphate.
12. The stable tablet formulation of claim 10 or 11, wherein the binder is
in
the range of about 1 wt% to about 5 wt%.
13. The stable tablet formulation of claim 12, wherein the binder is in the

range of about 1.5 wt% to about 3 wt%.
14. The stable tablet formulation of any one of claims 10 to 13, wherein
the
weight ratio of the binder to the tetrahydrobiopterin is in the range of about
1:10 to about
1:20.
15. The stable tablet formulation of claim 14, wherein the weight ratio of
the
binder to the tetrahydrobiopterin is about 1:15.

- 38 -
16. The stable tablet formulation of any one of claims 1 to 15, further
comprising a disintegration agent.
17. The stable table formulation of claim 16, wherein the disintegration
agent
is crospovidone.
18. The stable tablet formulation of claim 16 or 17, wherein the
disintegration
agent is in the range of about 3 wt% to about 10 wt%.
19. The stable tablet formulation of claim 18, wherein the disintegration
agent
is in the range of about 3 wt% to about 5 wt%.
20. The stable tablet formulation of any one of claims 16 to 19, wherein
the
weight ratio of the disintegration agent to the tetrahydrobiopterin is in the
range of about
1:5 to about 1:10.
21. The stable tablet formulation of claim 20, wherein the weight ratio of
the
disintegration agent to the tetrahydrobiopterin is about 1:7.5.
22. The stable tablet formulation of any one of claims 1 to 21, wherein the

antioxidant is an acidic antioxidant.
23. The stable tablet formulation of claim 22, wherein the acidic
antioxidant is
ascorbic acid.
24. The stable tablet formulation of claim 22 or 23, wherein the acidic
antioxidant is in the range of about 1 wt% to about 3 wt%.
25. The stable tablet formulation of any one of claims 22 to 24, wherein
the
weight ratio of the acidic antioxidant to the tetrahydrobiopterin is about
1:20.
26. The stable tablet formulation of any one of claims 1 to 25, further
comprising a lubricant.

- 39 -
27. The stable table formulation of claim 26, wherein the lubricant is
stearyl
fumarate.
28. The stable tablet formulation of claim 26 or 27, wherein the lubricant
is in
the range of about 0.1 wt% to about 2 wt%.
29. The stable tablet formulation of claim 28, wherein the lubricant is in
the
range of about 0.5 wt% to about 1.5 wt%.
30. The stable tablet formulation of any one of claims 26 to 29, wherein
the
weight ratio of the lubricant to the tetrahydrobiopterin is in the range of
about 1:25 to
about 1:65.
31. The stable tablet formulation of claim 30, wherein the weight ratio of
the
lubricant to the tetrahydrobiopterin is about 1:27.
32. The stable tablet formulation of any one of claims 1 to 31, further
comprising vitamin B2 (riboflavin).
33. The stable tablet formulation of any one of claims 1 to 32, further
comprising vitamin B12.
34. The stable tablet formulation of any one of claims 1 to 33, further
comprising a folate.
35. The stable tablet formulation of claim 34, wherein the folate is folic
acid
(pteroylmonoglutamate), dihydrofolic acid, tetrahydrofolic acid, 5-
methyltetrahydrofolic
acid, 5,10-methylenetetrahydrofolic acid, 5,10-methenyltetrahydrofolic acid,
5,10-formiminotetrahydrofolic acid, 5-formyltetrahydrofolic acid (leucovorin),

10-formyltetrahydrofolic acid, 10-methyltetrahydrofolic acid, a
folylpolyglutamate, a
dihydrofolate, a tetrahydrofolate, 5-formyl-(6S)-tetrahydrofolic acid, 5-
methyl-(65)-
tetrahydrofolic acid, 5,10-methylene-(6R)-tetrahydrofolic acid, 5,10-methenyl-
(6R)-
tetrahydrofolic acid, 10-formyl-(6R)-tetrahydrofolic acid, 5-formimino-(65)-

- 40 -
tetrahydrofolic acid or (6S)-tetrahydrofolic acid, or a pharmaceutically
acceptable salt
thereof.
36. The stable tablet formulation of any one of claims 1 to 35, further
comprising arginine.
37. A method of making the stable tablet formulation of any one of claims 1

to 36, comprising the steps of mixing an initial amount of said crystalline
polymorph of
(6R)-L-erythro-tetrahydrobiopterin and one or more pharmaceutically acceptable

excipients, and forming a tablet from the mixture, wherein the steps do not
include adding
liquid water.
38. The method of claim 37, wherein the one or more pharmaceutically
acceptable excipients include a binder.
39. The method of claim 38, wherein the binder is anhydrous dibasic calcium

phosphate.
40. The method of any one of claims 37 to 39, wherein the one or more
pharmaceutically acceptable excipients include a disintegration agent.
41. The method of claim 40, wherein the disintegration agent is
crospovidone.
42. The method of any one of claims 37 to 41, wherein the one or more
pharmaceutically acceptable excipients include an acidic antioxidant.
43. The method of claim 42, wherein the acidic antioxidant is ascorbic
acid.
44. The method of any one of claims 37 to 43, wherein the one or more
pharmaceutically acceptable excipients include a lubricant.
45. The method of claim 44, wherein the lubricant is stearyl fumarate.

- 41 -
46. A formulation, comprising about 100 mg of crystalline polymorph B of
(6R)-L-erythro-tetrahydrobiopterin dihydrochloride and about 5 mg of ascorbic
acid in
the form of a tablet;
wherein, after six months at room temperature and about 60% humidity, the
formulation retains at least about 95% of the initial amount of (6R)-L-erythro-

tetrahydrobiopterin; and
wherein the crystalline polymorph exhibits an X-ray powder diffraction pattern

with the following characteristic peaks expressed in d-values(.ANG.): 8.7
(vs), 5.63 (m),
4.76(m), 4.40 (m), 4.00 (s), 3.23 (s), and 3.11 (vs).
47. The formulation of claim 46, further comprising crospovidone, dibasic
calcium phosphate, D-mannitol, riboflavin, and sodium stearyl fumarate.
48. The formulation of claim 46, further comprising crospovidone.
49. The formulation of claim 46 or 48, further comprising dibasic calcium
phosphate.
50. The formulation of any one of claims 46, 48, and 49, further comprising

D-mannitol.
51. The formulation of any one of claims 46 and 48 to 50, further
comprising
riboflavin.
52. The formulation of any one of claims 46 and 48 to 51, further
comprising
sodium stearyl fumarate.
53. A formulation, comprising crystalline polymorph B of (6R)-L-erythro-
tetrahydrobiopterin dihydrochloride and ascorbic acid in the form of a tablet,
wherein the
weight ratio of ascorbic acid to (6R)-L-erythro-tetrahydrobiopterin is about
1:5.5, about
1:6, about 1:6.5, about 1:7, about 1:7.5, about 1:8, about 1:8.5, about 1:9,
about 1:9.5,
about 1:10, about 1:10.5, about 1:11, about 1:11.5, about 1:12, about 1:12.5,
about 1:13,
about 1:13.5, about 1:14, about 1:14.5, about 1:15, about 1:15.5, about 1:16,
about 1:16.5,

- 42 -
about 1:17, about 1:17.5, about 1:18, about 1:18.5, about 1:19, about 1:19.5,
about 1:20,
about 1:20.5, about 1:21, about 1:21.5, about 1:22, about 1:22.5, about 1:23,
about 1:23.5,
about 1:24, about 1:24.5, about 1:25, about 1:25.5, about 1:26, about 1:26.5,
about 1:27,
about 1:27.5, about 1:28, about 1:28.5, about 1:29, or about 1:29.5.
54. The formulation of claim 53, further comprising crospovidone, dibasic
calcium phosphate, D-mannitol, riboflavin, and sodium stearyl fumarate.
55. The formulation of claim 53, further comprising crospovidone.
56. The formulation of claim 53 or 55, further comprising dibasic calcium
phosphate.
57. The formulation of any one of claims 53, 55, and 56, further comprising

D-mannitol.
58. The formulation of any one of claims 53 and 55 to 57, further
comprising
riboflavin.
59. The formulation of any one of claims 53 and 55 to 58, further
comprising
sodium stearyl fumarate.
60. The formulation of any one of claims 53 to 59, wherein the weight ratio
of
ascorbic acid to (6R)-L-erythro-tetrahydrobiopterin is about 1:16.5, about
1:17, about
1:17.5, about 1:18, about 1:18.5, about 1:19, about 1:19.5, about 1:20, about
1:20.5, about
1:21, about 1:21.5, about 1:22, about 1:22.5, about 1:23, about 1:23.5, about
1:24, about
1:24.5, or about 1:25.
61. The formulation of claim 60, wherein the weight ratio of ascorbic acid
to
(6R)-L-erythro-tetrahydrobiopterin is about 1:19, about 1:19.5, or about 1:20.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02581814 2012-11-02
-1-
STABLE TABLET FORMULATION OF TETRAHYDROBIOPTERIN
BACKGROUND
Field
The present invention is generally directed to stable tablet formulations
of tetrahydrobiopterin or precursors, derivatives or analogs thereof for the
treatment
of humans.
Background of the Related Technology
Tetrahydrobiopterin (sometimes referred to as BH4) is a biogenic
amine of the naturally-occurring pterin family that is a cofactor for a number
of
different enzymes, including phenylalanine hydroxylase (PAH), tyrosine
hydroxylase,
tryptophan hydroxylase and nitric oxide synthase. Pteiins are present in
physiological
fluids and tissues in reduced and oxidized forms, however, only the 5,6,7,8,
tetrahydrobiopterin is biologically active. It is a chiral molecule and the 6R

enantiomer of the cofactor is known to be the biologically active enantiomer.
For a
detailed review of the synthesis and disorders of BH4 see Blau et al., 20W
(Disorders
of tetrahydrobioptertn ond related biogenic amines. In: Scriver CR, Beaudet
AL, Sly
WS, Valle D, Chi!es 3, Vogelstejn B, eds. The Metabolic and Molecular Bases of

inherited Distase. 8th ed. New Yolk: McGraw-Hill, 2001: 1275-1776).
A deficiency in PAH activity, due to absent or mutated PAH enzyme
or a deficiency in its cofactor BH4, manifests as an excess of the amino acid
phenylalanine (Phe) known as hyperphenylalaninemia (1-IPA) in its very mildest

forms or phenylketonuria (PKU) in its moderate or severe forms. PAH deficiency

also causes a deficiency in the amino acid tyrosine, which is a precursor for
synthesis
of neurotransmitters. A deficiency in tyrosine hydroxylase or tryptophan
hydroxylase
activity can manifest as a deficiency in neurotransmitter production.
Despite the elucidation of the role of BH4 deficiency in
phenylketonuria, treatment with BH4 has not been suggested because such
treatment
is very expensive, as high as $30,000 per year for an adolescent or adult, as
compared
with $6,000 for phenylalanine-restricted dietary therapy (Hanley, N. EngL J.
Med
348(17):1723, 2003). Another significant problem with BH4 is that this
compound is
unstable and readily undergoes aerobic oxidation at room temperature (Davis et
al.,
Eur. J. Biochem., Vol 173, 345-351, 1988; U.S. Patent No. 4,701,455) and has a

CA 02581814 2007-03-22
WO 2006/055511
PCT/US2005/041252
-2-
shelf-life of less 8 hours at room temperature (Berneggar and Blau, Mol.
Genet.
Metabol. 77:304-313, 2002).
Other tetrahydrobiopterin products available on the market need to be
specially packaged or kept frozen. For example the labeling on the tablets
sold by
Schirck's Laboratory specify that the tablets should be kept frozen and state
that the
product has a shelf life at room temperature of only 2 months. BIOPTEN
(tetrahydrobiopterin granules) requires expensive, hermetically-sealed foil
packaging
to maintain room temperature stability. The instability of such BH4
compositions is
commercially undesirable and significant degradation due to improper storage
could
hinder therapy of patients.
Drug substance polymorphic forms can exhibit different physical and
mechanical properties, including hygroscopicity, particle shape, density,
flowability,
and compactibility, which in turn may affect processing of the drug substance
and/or
manufacturing of the drug product. The effect of polymorphism on
pharmaceutical
processing also depends on the formulation and the manufacturing process.
Polymorphic forms of the drug substance can undergo phase conversion when
exposed to a range of manufacturing processes, such as drying, milling,
micronization, wet granulation, spray-drying, and compaction. Exposure to
environmental conditions such as humidity and temperature can also induce
polymorph conversion. The extent of conversion generally depends on the
relative
stability of the polymorphs, kinetic barriers for phase conversion, and
applied stress.
See FDA Center for Drug Evaluation and Research (CDER) Draft Guidance for
Industry ANDAs: Pharmaceutical Solid Polymorphism Chemistry, Manufacturing,
and Controls Information, December 2004.
Thus, there remains a need for a stable solid formulation of
tetrahydrobiopterin and processes for manufacturing such stable formulations.
The
present invention is directed to addressing such a need.

CA 02581814 2014-07-30
a
¨ 3 -
SUMMARY OF THE INVENTION
Certain exemplary embodiments provide a stable tablet formulation
comprising an initial amount of a crystalline polymorph, designated polymorph
B, of
(6R)-L-erythro-tetrahydrobiopterin, an antioxidant, and a pharmaceutically
acceptable
excipient, wherein the weight ratio of the antioxidant to (6R)-L-erythro-
tetrahydrobiopterin is in the range of about 1:5 to about 1:30; wherein, after
six months at
room temperature and about 60% humidity, the stable tablet formulation retains
at least
about 95% of the initial amount of (6R)-L-erythro-tetrahydrobiopterin; and
wherein said
crystalline polymorph, as a hydrochloride salt, exhibits an X-ray powder
diffraction
pattern with the following characteristic peaks expressed in d-values(A): 8.7
(vs),
5.63 (m), 4.76(m), 4.40 (m), 4.00 (s), 3.23 (s), and 3.11 (vs).
Further certain exemplary embodiments provide a formulation, comprising
about 100 mg of crystalline polymorph B of (6R)-L-erythro-tetrahydrobiopterin
dihydrochloride and about 5 mg of ascorbic acid in the form of a tablet;
wherein, after six
months at room temperature and about 60% humidity, the formulation retains at
least
about 95% of the initial amount of (6R)-L-erythro-tetrahydrobiopterin; and
wherein the
crystalline polymorph exhibits an X-ray powder diffraction pattern with the
following
characteristic peaks expressed in d-values(A): 8.7 (vs), 5.63 (m), 4.76(m),
4.40 (m),
4.00 (s), 3.23 (s), and 3.11 (vs).
Further certain exemplary embodiments provide a formulation, comprising
crystalline polymorph B of (6R)-L-erythro-tetrahydrobiopterin dihydrochloride
and
ascorbic acid in the form of a tablet, wherein the weight ratio of ascorbic
acid to (6R)-L-
erythro-tetrahydrobiopterin is about 1:5.5, about 1:6, about 1:6.5, about 1:7,
about 1:7.5,
about 1:8, about 1:8.5, about 1:9, about 1:9.5, about 1:10, about 1:10.5,
about 1:11, about
1:11.5, about 1:12, about 1:12.5, about 1:13, about 1:13.5, about 1:14, about
1:14.5, about
1:15, about 1:15.5, about 1:16, about 1:16.5, about 1:17, about 1:17.5, about
1:18, about
1:18.5, about 1:19, about 1:19.5, about 1:20, about 1:20.5, about 1:21, about
1:21.5, about
1:22, about 1:22.5, about 1:23, about 1:23.5, about 1:24, about 1:24.5, about
1:25, about
1:25.5, about 1:26, about 1:26.5, about 1:27, about 1:27.5, about 1:28, about
1:28.5, about
1:29, or about 1:29.5.

CA 02581814 2012-11-02
-3a-
The present invention relates to stable solid formulations of
tetrahydrobiopterin, particularly stable tablets, processes for producing such
formulations, and treatment methods using such formulations.
The invention provides a stable solid formulation of
tetrahydrobiopterin, or a precursor or derivative or analog thereof, that
maintains its
stability for an extended period of time. Compositions of the invention may
comprise
a stable, crystalline form of BH4 that is stable at room temperature for more
than 8
hours and a pharmaceutically acceptable carrier, diluent or excipient.
Exemplary
stable tablets of the invention have been prepared using a dry tableting
process and
have been shown to have a shelf-life of at least 6 to 9 months at room
temperature.
Another aspect of the invention provides a dry formulation process for
preparing stable solid formulations, which includes the step of mixing
tetrahydrobiopterin, or a precursor or derivative or analog thereof, with
another
pharmaceutical carrier, diluent or excipient, in the absence of added water.
In an exemplary embodiment, the active pharmaceutical ingredient and
excipients are dry blended and compressed. The tablets are processed in
humidity-
controlled rooms where humidity is kept at about 65% (- 5%) or less. Once
processed, the tablets are stored in triple plastic lined water resistant
containers with
desiccant pillows lined between the outer two layers of plastic hags. Thus,
the
invention includes a dry formulation method comprising the steps of mixing an
initial
amount of a crystalline polymorph of (6R)-L---erythro-tetrahydrobiopterin,
preferably
polymorph B, and one or more pharmaceutically acceptable excipients, and
forming a
tablet from the mixture, wherein the steps do not include adding liquid water.
Exemplary particle sizes include, e.g., from about 0.2 jim to about 500 gm,
from
about 1 gm to about 250 gm, or from about 2 gm to about 200 gm, or, e.g.,
larger than
about 500 gm, larger than about 600 pm, larger than about 700 gm, or smaller
than
about 850 gm.
In exemplary embodiments, the tablet is initially manufactured using a
stable crystalline form of (6R)-5,6,7,8-tetrahydrobiopterin described below as

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-4-
"polymorph B," and retains at least about 95% of the active pharmaceutical
ingredient
(API) at room temperature after 3 months, 6 months or 9 months, or preferably
12
months or longer, e.g. 15 months, 18 months, 21 months, 2 years, 2.5 years, 3
years or
longer. Preferably the tablet retains at least about 90%, 91%, 92%, 93%, 94%,
95%,
96%, 97%, 98%, 99%, or 99.5% of the API after storage at room temperature for
such
time periods. The tablet also preferably exhibits loss on drying of 2% or
less, or 1.5%
or less, or 1% or less, or 0.9% or less, or 0.8% or less, or 0.7% or less, or
0.6% or less,
after such time periods. Exemplary tablets may be manufactured wherein the
initial
amount of tetrahydrobiopterin active pharmaceutical ingredient is about 25 mg,
50
mg, 75 mg, 100 mg, 125 mg, 150 mg, 175 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400
mg, 450 mg, 500 mg, 550 mg, 600 mg, 650 mg, 700 mg, 750 mg, 800 mg, 850 mg,
900 mg or higher doses. Preferred tablets also exhibit rapid disintegration
upon
administration, e.g., 3 minutes or less, to improve ease of administration.
Thus, the invention provides a stable tablet formulation comprising an
initial amount of a crystalline polymorph, designated polymorph B, of (6R)-L-
erythro-tetrahydrobiopterin and a pharmaceutically acceptable excipient,
wherein
after six months at room temperature and about 60% humidity the stable tablet
formulation retains at least about 95% of the initial amount of (612)-L-
erythro-
tetrahydrobiopterin, and wherein said crystalline polymorph, as a
hydrochloride salt,
exhibits an X-ray powder diffraction pattern with the following characteristic
peaks
expressed in d-values(A) : 8.7 (vs), 5.63 (m), 4.76(m), 4.40 (m), 4.00 (s),
3.23 (s),
3.11 (vs), preferably 8.7 (vs), 6.9 (w), 5.90 (vw), 5.63 (m), 5.07 (m), 4.76
(m), 4.40
(m), 4.15 (w), 4.00(s), 3.95 (m), 3.52 (m), 3.44 (w), 3.32 (m), 3.23 (s), 3.17
(w), 3.11
(vs), 3.06 (w), 2.99 (w), 2.96 (w), 2.94 (m), 2.87 (w), 2.84 (s), 2.82 (m),
2.69 (w),
2.59 (w), and 2.44 (w). Preferably the tablet retains at least about 90%, 91%,
92%,
93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.5% of the initial amount of (6R)-L-
erythro-tetrahydrobiopterin.
The stable solid formulation preferably contains one or more of the
following additional ingredients that improve stability or other
characteristics of the
formulation: binder, disintegrant, acidic antioxidant, or lubricant or
combinations
thereof. One exemplary preferred composition includes anhydrous dibasic
calcium
phosphate, crospovidone, ascorbic acid and stearyl fumarate, optionally with
mannitol

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-5-
and riboflavin. The stable solid formulation may optionally include other
therapeutic
agents suitable for the condition to be treated, e.g. folates, including
folate precursors,
folic acids, or folate derivatives; and/or vitamins such as vitamin C and/or
vitamin
B12; and/or neurotransmitter precursors such as L-dopa or carbidopa; and/or 5-
hydroxytryptophan; and/or arginine. Compositions comprising
tetrahydrobiopterin
(or a precursor or derivative or analog) and a folate, and optionally further
comprising
arginine, are particularly contemplated.
The invention further contemplates other stable solid formulations for
oral administration, e.g. capsules, pills or troches, with similar stability
properties.
Yet another.aspect of the invention provides treatment methods using
such stable solid formulations. The invention contemplates that such
formulations of
the invention are useful for intervention in metabolic disorders, particularly
those
involving amino acid metabolism. More particularly, the stable formulations
may be
used for the treatment of subjects exhibiting elevated phenylalanine levels or
decreased tyrosine levels, for example, subjects suffering from
hyperphenylalanemia,
mild phenylketonuria or classic severe phenylketonuria: and far the treatment
of
subjects suffering from conditions that would benefit from enhancenient of
nitric
oxide synthase activity, including vascular diseases, ischemic or inflammatory

diseases, diabetes, or insulin resistance. The total dose required for each
treatment
may be administered in multiple doses or in a single dose. The stable
formulations
may be administered daily or at some other interval, e.g., every alternative
day or
even weekly.
The stable formulations may be used alone or in conjunction with other
therapies suitable for the disorder to be treated, including the underlying
disease or the
clinical symptoms. For example, for HPA, the stable formulations of the
invention
may be administered in combination with a protein-restricted diet, e.g. where
the
subject is limited to about 600 mg or less, or about 300 mg or less of protein
daily,
optionally with supplemental amino acids, such as tyrosine, valine, isoleucine
and
leucine. The stable formulations may also be administered in combination with
folates, arginine, vitamins, or neurotransmitter precursors. As another
example, for
vascular diseases, diabetes, or insulin resistance, the stable formulations of
the
invention may be administered in conjunction with other therapeutic agents
such as

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-6-
anti-hypertensive agents, anti-platelet agents, cholesterol-lowering agents,
insulin or
oral hypoglycemic agents.
Other features and advantages of the invention will become apparent
from the following detailed description. It should be understood, however,
that the
detailed description and the specific examples, while indicating preferred
embodiments of the invention, are given by way of illustration only, because
various
changes and modifications within the spirit and scope of the invention will
become
apparent to those skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows a characteristic X-ray powder diffraction pattern for
crystalline polymorph B of (6R)-5,6,7,8-tetrahydrobiopterin dihydrochloride.
DESCRIPTION OF THE PREFERRED EMBODIMENTS
=
The present invention provides a stable formulation that Maintains a
stable crystalline polymorph of the active ingredient. An anhydrous polymorph
of
(6R)-5,6,7,8-tetrahydrobiopterin dilrydrochloride that is stable at room
temperature to
atmospheric oxygen and normal humidity, described below as polymorph B, has
been
identified. However, when the percent relative humidity approaches 80%,
polymorph
B appears to take up much more water, loses its crystalline form, and becomes
labile
to oxidation.
By using a dry formulation process, the stable crystalline structure of
this polymorph is maintained in the finished product. In contrast, other
processes for
preparing tetrahydrobiopterin compositions result in a less stable product
compared to
those of the present invention.
Stable tablet formulations of the invention have been made using
polymorph B in a dry formulation process and have been shown to retain 99% or
more of the initial (6R)-5,6,7,8-tetrahydrobiopterin for at least 6 or 9
months, both at
normal room temperature and humidity, and under accelerated testing
conditions.
The observed stability under accelerated testing conditions, i.e. higher
temperature

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-7-
and humidity, indicates that the tablet formulations would be stable for far
longer than
6 or 9 months at normal room temperature and humidity.
As used herein, "shelf life" means the storage period during which an
active pharmaceutical ingredient (API) in a pharmaceutical formulation has
minimal
degradation (e.g., not more than about 5% degradation) when the pharmaceutical
formulation is stored under specified storage conditions, for example, room
temperature at normal humidity.
The shelf-life of the stable formulations of the invention may be
measured as follows. The formulation to be tested may be divided into one or
more
different batches and stored under typical storage conditions, for example, 4
C
(refrigerator), or 25 C (room temperature). Degradation of the API in a
pharmaceutical formulation can also be detected using accelerated testing
under
exaggerated storage conditions designed to increase the degradation rate of
the drug
substance. For example, a batch can be "stressed" (placed in chamber which
maintains a temperature of 45 C and 75% humidity). Samples of each batch of
formulation are then analyzed at different time points (e.g., time zero,,2
weeks, 1
month, 3 months, 6 months, 9 months, 1 year, [.5 years, 2.years, 2.5 years, 3
years or
longer) for amount of API still present in the formulation. Analysis of the
API in the
formulation may be carried out by a variety of detection methods including
high
performance liquid chromatography, crystal or powder X-ray diffraction,
infrared or
Raman spectra studies, microscopy, differential scanning calorimetry, thermal
gravimetric analysis, hot-stage microscopy, and solid state nuclear magnetic
resonance. Maintenance of a particular polymorph form can be determined by
carrying out, e.g., powder or crystal X-ray diffraction studies or any of the
same
techniques used to analyze the polymorph initially.
I. Synthesis of tetrahydrobiopterin, precursors, derivatives and
analogs
A variety of methods are known in the art for synthesis of
tetrahydrobiopterins, precursors, derivatives and analogs. U.S. Patent Nos.
5,698,408;
2,601,215; 3505329; 4,540,783; 4,550,109; 4,587,340; 4,595,752; 4,649,197;
4,665,182; 4,701,455; 4,713,454; 4,937,342; 5,037,981; 5,198,547; 5,350,851;

CA 02581814 2010-11-10
-8-
5,401,844; 5,698,408, Canadian application CA 2420374, European application
nos.
EP 079 574, EP 191 335 and Suntory Japanese patent publications JP 4-082888,
JP
59-021685 and JP 9-157270, as well as Sugimoto and Matsuura, Bull. Chem. Soc.
Japan, 48(12):3767-3768 (1975), Sugimoto and Matsuura, Bull. Chem. Soc. Japan,
52(1):181-183 (1979), Matsuura etal., Chem. Lett. (Japan), 735-738 (1984),
Matsuura etal., Heterocycles, Vol. 23, No. 12, 3115-3120, 1985 and Whiteley
etal.,
Anal Biochem. 137(2):394-6 (1984) each describe methods of making
dihydrobiopterins, BH4 and derivatives thereof that may be used as
compositions for the present invention.
Int'l Publication No. W02005049614, U.S. Patent No. 4,540,783,
Japanese Patent No. 59-021685, Schircks et al., Hely. Chim. Acta, 60: 211
(1977),
Sugimoto et al., Bull. Chem. Soc. Jp, 52(1):181 (1979), Sugimoto et al., Bull.
Chem.
Soc. Jp, 48(12):3767 (1975), Visontini etal., Hely. Chim. Acta, 52:1225
(1969), and
Matsuura et al., Chem. Lett., p 735 (1984) describe methods of synthesizing
BH4.
Nonlimiting examples of analogs for use in the compositions and =
methods described herein include pteridine, pterin, neopterin, biopterin, 7,8-
Dihydrobiopterin, 6-methyltetrahydropterin, and other 6-substituted
tetrahydropterin
and other 6-substituted tetrahydropterins, sepiapterin, 6,7-
Dimethyltetrahydropterin,
6-methyl biopterin and other 6-substituted biopterins, and other analogs that
are
described in the art. Nonlimiting examples of derivatives for use in the
compositions
and methods described herein include the derivatives described in U.S. Patent
Nos.
4,758,571; 4,774,244; 6,162,806; 5,902, 810; 2,955,110; 2,541,717; 2,603,643;
and
4,371,514.
Any such methods or other methods known in the art may be used to
produce BH4, or precursors, derivatives or analogs for use in the stable
formulations
and therapeutic methods of the present invention.

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-9-
II. Crystal Polymorphs of 6R-tetrahydrobiopterin hydrochloride salt
It has been found that BH4, and in particular, the dihydrochloride salt
of BH4, exhibits crystal polymorphism. The structure of BH4 is shown below:
0 OH
H
HN/ N
a
H2N
The (6R) form of B114 is the known biologically active form, however, BH4 is
known
to be unstable at ambient temperatures.
BH4 is difficult to handle and it was therefore produced and offered as
its dihydrochloride salt (Schircks Laboratories, Jona, Switzerland) in
ampoules sealed
under nitrogen to prevent degradation of the substance due to its hygroscopic
nature
and sensitivity to oxidation. U.S. Patent No. 4,649,197 discloses that
separation of
(6R)- and 6(S)-L-erythro-tetrahydrobiopterin dihydrochloride into its
diastereomers is
difficult due to the poor crystallinity of 6(R,S)-L-orythro-
tetrahydrobiopterin
dihydrochlcride. The European patent Dumber 0 079 574 describes the
preparation of
tetrahydrobiopterin, wherein a solid tetrahydrobiopterin dihydrochloride is
obtained
as an intermediate. S. Matsuura et al. describes in Chemistry Letters 1984,
pages 735-
738 and Heterocycles, Vol. 23, No. 12, 1985 pages 3115-3120 6(R)-
tetrahydrobiopterin dihydrochloride as a crystalline solid in form of
colorless needles,
which are characterized by X-ray analysis disclosed in J. Biochem. 98, 1341-
1348
(1985). An optical rotation of 6.81 was found the crystalline product, which
is quite
similar to the optical rotation of 6.51 reported for a crystalline solid in
form of white
crystals in example 6 of EP-A2-0 191 335.
Results obtained during development of (6R)-L-erythro-
.
tetrahydrobiopterin dihydrochloride indicated that the compound may exist in
different crystalline forms, including polymorphic forms and solvates. It has
been
found that one crystal polymorph of BH4 is more stable, and is stable to
decomposition under ambient conditions.

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-10-
Polymorph Form B
The crystal polymorph that has been found to be the most stable is
referred to herein as "form B," or alternatively as "polymorph B."
Polymorph B is a slightly hygroscopic anhydrate with the highest
thermodynamic stability above about 20 C. Furthermore, form B can be easily
processed and handled due to its thermal stability, possibility for
preparation by
targeted conditions, its suitable morphology and particle size. Melting point
is near
260 C (AHf > 140 J/g), but no clear melting point can be detected due to
decomposition prior and during melting. These outstanding properties renders
polymorph form B especially feasible for pharmaceutical applications, which
are
often prepared at elevated temperatures. Polymorph B can be obtained as a fine

powder with a particle size that may range from 0.2 1.1M to 500 pm.
Form B exhibits an X-ray powder diffraction pattern, expressed in d-
values (A) at: 8.7 (vs), 6.9 (w), 5.90 (vw), 5.63 (m), 5.07 (m), 4.76 (m),
4.40 (m), 4.15
(w), 4.90 (s), 3.95 (m), 3.52 (m), 3.44 (w),3.32 (m), 3.23 (s), 3.17 (w), 3.11
(vs), 106
(w), 2.99 (w), 2.96 (w), 2.94 (m), 2.87 (w), 2.84 (s), 2.82 (m), 2.69 (w),2.59
(w), 2'.44
(w). Figure 1 is a graph of the characteristic X-ray diffraction pattern
exhibited by
form B of (6R)-L-erythro-tetrahydrobiopterin dihydrochloride. As used herein,
the
=
following the abbreviations in brackets mean: (vs) = very strong intensity;
(s) = strong
intensity; (m) = medium intensity; (w) = weak intensity; and (vw) = very weak
intensity.
Form B can be prepared in very large quantities (e.g., 100 kilo scale)
and stored over an extended period of time.
All crystal forms (polymorphs, hydrates and solvates), inclusive crystal
form B, can be used for the preparation of the most stable polymorph B.
Polymorph
B may be obtained by phase equilibration of suspensions of amorphous or other
forms, in suitable polar and non aqueous solvents.
Other forms of BH4 can be converted into form B by dispersing the
other form of BH4 in a solvent at room temperature, stirring the suspension at
ambient temperatures for a time sufficient to produce polymorph form B,
thereafter
isolating crystalline form B and removing the solvent from the isolated form
B.

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-11-
Ambient temperatures, as used herein, mean temperatures in a range from 0 C
to 60
C, preferably 15 C to 40 C. The applied temperature may be changed during
treatment and stirring by decreasing the temperature stepwise or continuously.

Suitable solvents for the conversion of other forms to form B include but are
not
limited to, methanol, ethanol, isopropanol, other C3- and C4-alcohols, acetic
acid,
acetonitrile, tetrahydrofurane, methy-t-butyl ether, 1,4-dioxane, ethyl
acetate,
isopropyl acetate, other C3-C6-acetates, methyl ethyl ketone and other methyl-
C3-05
alkyl-ketones. The time to complete phase equilibration may be up to 30 hours
and
preferably up to 20 hours or less than 20 hours.
Polymorph B may also be obtained by crystallisation from solvent
mixtures containing up to about 5% water, especially from mixtures of ethanol,
acetic
acid and water. It has been found that polymorph form B of (6R)-L-erythro-
tetrahydrobiopterin dihydrochloride can be prepared by dissolution, optionally
at
elevated temperatures, preferably of a solid lower energy form than form B or
of form
B of (6R)-L-erythro-tetrahydrobiopterin dihydrochloride in a solvent mixture
comprising ethanol, acetic acid and water, addition of seeds to the aqution,
cooling
the obtained suspension and isolation of the formed crystals. Dissol GI ion
may be
carried out at room temperature or up to 70 C, preferably up to 50 C. There
may be
used the final solvent mixture for dissolution or the starting material may be
first
dissolved in water and the other solvents may than be added both or one after
the
other solvent. The composition of the solvent mixture may comprise a volume
ratio of
water: acetic acid : tetrahydrofuran of 1 : 3: 2 to 1: 9: 4 and preferably 1:
5: 4. The
solution is preferably stirred. Cooling may mean temperatures down to -40 C
to 0 C,
preferably down to 10 C to 30 C. Suitable seeds are polymorph form B from
another batch or crystals having a similar or identical morphology. After
isolation, the
crystalline form B can be washed with a non-solvent such as acetone or
tetrahydrofurane and dried in usual manner.
Polymorph B may also be obtained by crystallization from aqueous
solutions through the addition of non-solvents such as methanol, ethanol and
acetic
acid. The crystallisation and isolation procedure can be advantageously
carried out at
room temperature without cooling the solution. This process is therefore very
suitable
to be carried out at an industrial scale.

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-12-
In one embodiment of the compositions and methods described herein,
a composition including polymorph form B of (6R)-L-erythro-tetrahydrobiopterin

dihydrochloride is prepared by dissolution of a solid form other than form B
or of
form B of (6R)-L-erythro-tetrahydrobiopterin dihydrochloride in water at
ambient
temperatures, adding a non-solvent in an amount sufficient to form a
suspension,
optionally stirring the suspension for a certain time, and thereafter
isolation of the
formed crystals. The composition is further modified into a pharmaceutical
composition as described below.
The concentration of (6R)-L-erythro-tetrahydrobiopterin
dihydrochloride in the aqueous solution may be from 10 to 80 percent by
weight,
more preferably. from 20 to 60 percent by weight, by reference to the
solution.
Preferred non-solvents (i.e., solvents useful in preparing suspensions of BH4)
are
methanol, ethanol and acetic acid. The non-solvent may be added to the aqueous

solution. More preferably, the aqueous solution is added to the non-solvent.
The
-15 stirring time after formation of the suspension may be up to 30 hours
and preferably
up to 20 hours or less than 20 hours. Isolation by filtration and dryiii:;iS
carried out in -
known manner as described above.
Polymorph form B is a very stable crystalline form, thai can be easily
filtered off, dried and ground to particle sizes desired for pharmaceutical
formulations. These outstanding properties renders polymorph form B especially
feasible for pharmaceutical application.
III. Stable Pharmaceutical Formulations
Pharmaceutical formulations may initially include a stable crystalline
form of tetrahydrobiopterin, or a precursor or derivative or analog thereof,
with a
pharmaceutically acceptable carrier. The stable formulation of the invention
preferably contains one or more of the following additional ingredients that
improve
the stability or other characteristics of the formulation: binder,
disintegration agent,
acidic antioxidant, or lubricant or combinations thereof. Preferably a stable
tablet
formulation includes a binder and disintegration agent, optionally with an
acidic
antioxidant, and optionally further including a lubricant.

CA 02581814 2010-11-10
-13-
The initial amount of a crystalline polymorph of (6R)-L-erythro-
tetrahydrobiopterin used to prepare the formulation may be, for example, in
the range
of about 30 wt% to about 40 wt% of the formulation, or in the range of about
32 wt%
to about 35 wt%, or at about 33 wt%.
Binders assist in maintaining the tablet formulation. In some cases,
anhydrous binders are used to preserve the anhydrous state of polymorph B. In
some
cases, the binder may act as a drying agent. Exemplary binders include
anhydrous
dibasic calcium phosphate and its monohydrate.
Exemplary concentrations of the binder in a stable tablet formulation
of the present invention are between about 1 wt% to about 5 wt%. Particularly
contemplated concentrations are between about 1.5 and 3 wt%. Also contemplated

are concentrations of binder of at least about 1.6, 1.7, 1.8, 1.9, 2.0,
2.1,2.2, 2.3, 2.4,
2.5, 2.6, 2.7, 2.8, 2.9, and 3.0 wt%, or concentrations up to about 3.1, 3.2,
3.3, 3.4,
3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, and
5.0 wt%. The
weight ratio of binder to tetrahydrobiopterin in a stable tablet formulation
of the
present invention is, for example, in the range of about 1:10 to about 1:20.
Also
contemplated are weight ratios of aboui. 1:10.25, 1:1G.5, 1:1O.7, 1:11,
1:11.25,
1:11.5, 1:11.75, 1:12, 1:12.25, 1:12.5,1:12.75, 1:13, 1:13.25, 1:13.5,
1:13.75,1:14,
1:14.25, 1:14.5, 1:14.75, 1:15, 1:15.25, 1:15.5, 1:15.75, 1:16, 1:16.25,
1:16.5,1:16.75,
1:17, 1:17.25, 1:17.5, 1:17.75, 1:18, 118.25, 1:18.5, 1:18.75, 1:19, 1:19.25,
1:19.5,
and 1:19.75.
Disintegration agents assist in rapid disintegration of solid
formulations by absorbing water and expanding. Exemplary disintegration agents

include polyvinylpyrrolidone (PVP, e.g. sold under the name POVIDONETm), a
cross-
linked form of povidone (CPVP, e.g. sold under the name CROSPOVIDONETm), a
cross-linked form of sodium carboxymethylcellulose (NaCMC, e.g. sold under the

name AC-DI-SOLTm), other modified celluloses, and modified starch. Tablets
formulated with CPVP exhibited much more rapid disintegration than tablets
formulated with PVP.
Exemplary concentrations of the disintegration agent in a stable table
formulation of the present invention are between about 1 wt% to about 20 wt%.

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-14-
Particularly contemplated concentrations are between about 3 wt% and about 10
wt%.
Also contemplated are concentrations of disintegration of at least about 1.1,
1.2, 1.3,
1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8,
2.9, and 3.0 wt%, or
concentrations up to about 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9,
5.0, 5.2, 5.4,
5.6, 5.7, 5.8, 6.0, 6.25, 6.5, 6.75, 8.0, 8.25, 8.5, 8.75, 9.0, 9.25, 9.5,
9.75, and 10.0
wt%. The weight ratio of disintegration agent to tetrahydrobiopterin in a
stable tablet
formulation of the present invention is, for example, in the range of about
1:5 to about
1:10. Also contemplated are weight ratios of about 1:5.25, 1:5.5, 1:5.75,
1:6.0,
1:6.25, 1:6.5, 1:6.75, 1:7.0, 1:7.25, 1:7.5, 1:7.75, 1:8.0, 1:8.25, 1:8.5,
1:8.75, 1:9.0,
1:9.25, 1:9.5, and 1:9.75.
Antioxidants may be included and help stabilize the
tetrahydrobiopterin product, especially after dissolution. Low pH aqueous
solutions
of API are more stable than are solutions at high pH. Exemplary acidic
antioxidants
include ascorbic acid, fatty acid esters of ascorbic acid such as ascorbyl
palmitate and
ascorbyl stearate, and salts of ascorbic acid such as sodium, calcium, or
potassium
ascorbate. Non-acidic antioxidants may also be usc n the stable tablet
formulations.
Noutiraiting examples of non-acidic antioxidants ciucly:ta-caroteTle,,alpha-
tocopherol. Acidic additives may be added to enhance stability of the tablet
formulation, including citric acid or malic acid.
Exemplary concentrations of the antioxidant in a stable table
formulation of the present invention are between about 1 wt% and about 3 wt%.
Particularly contemplated concentrations are at least about 1.1, 1.2, 1.3,
1.4, 1.5, 1.6,
1.7, 1.8, 1.9, and 2.0 wt%, or concentrations up to about 2.1, 2.2, 2.3, 2.4,
2.5, 2.6,
2.7, 2.8, 2.9, and 3.0 wt%. The weight ratio of antioxidant to
tetrahydrobiopterin in a
stable tablet formulation of the present invention is, for example, in the
range of about
1:5 to 1:30. Also contemplated are weight ratios of about 1:5.5, 1:6, 1:6.5,
1:7, 1:7.5,
1:8, 1:8.5, 1:9, 1:9.5, 1:10, 1:10.5, 1:11, 1:11.5, 1:12, 1:12.5, 1:13,
1:13.5, 1:14,
=
1:14.5, 1:15, 1:15.5, 1:16, 1:16.5, 1:17, 1:17.5, 1:18,1:18.5, 1:19, 1:19.5,
1:20, 1:20.5,
1:21, 1:21.5, 1:22, 1:22.5, 1:23, 1:23.5, 1:24, 1:24.5, 1:25, 1:25.5, 1:26,
1:26.5, 1:27,
1:27.5, 1:28, 1:28.5, 1:29, and 1:29.5.
In Schirk's Laboratory tablets, ascorbic acid is present at a ratio of 1:1
to BH4. In contrast, the concentration of ascorbic acid in the stable
formulations of

CA 02581814 2010-11-10
-15-
the invention is far lower, e.g. a weight ratio (mg/mg) of 1:20 of ascorbic
acid to BH4.
Thus, the invention also contemplates formulations comprising ascorbic acid at
a ratio
to BH4 of less than 1:1, e.g. 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10,
1:11, 1:12, 1:13,
1:14, 1:15, 1:16, 1:17, 1:18 or 1:19, preferably a ratio of less than 1:10.
Lubricants improve stability, hardness and uniformity of solid
formulations. Exemplary lubricants include stearyl fumarate and magnesium
stearate.
Exemplary concentrations of the lubricant in a stable tablet formulation
of the present invention are between about 0.1 wt% and about 2 wt%.
Particularly
contemplated concentrations are between about 0.5 and 1 wt%. Also contemplated
are concentrations of lubricant of at least about 0.1, 0.2, 0.3, 0.4, 0.5,
0.6, 0.7, 0.8, 0.9,
and 1.0 wt%, or concentrations up to about 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7,
1.8, 1.9,
and 2.0 wt%. The weight ratio of lubricant to tetrahydrobiopterin in a stable
tablet
formulation of the present invention is, for example, in the range of about
1:25 to
1:65. Also contemplated are weight ratios of about 1:26, 1:27, 1:28, 1:29,
1:30, 1:31,
1:32, 1:33, 1:34, 1:35, 1:36, 1:37, 1:38; 1:39, 1:40, 1:41, 1:42, 1:43, i:44.
1:45,1:46,
,
1:48, 1:49, 1:50, 1:51, 1:52, 1:53, 1:54, 155, i :56, 1:57, t :59,
!:'60, 1:61,
1:62, 1:63, 1:64, and 1:65.
The stable solid formulation may optionally include other therapeutic
agents suitable for the condition to be treated, e.g. folates, including
folate precursors,
folic acids, or folate derivatives; and/or arginine; and/or vitamins, such as
vitamin C
and/or vitamin B2 (riboflavin) and/or Vitamin B12; and/or neurotransmitter
precursors
such as L-dopa or carbidopa; and/or 5-hydroxytryptophan.
Exemplary folates, including folate precursors, folic acids, or folate
derivatives, are disclosed in U.S. Patent Nos. 6,011,040 and 6,544,994,
and include folic acid (pteroylmonoglutamate),
dihydrofolic acid, tetrahydrofolic acid, 5-methyltetrahydrofolic acid, 5,10-
methylenetetrahydrofolic acid, 5,10-methenyltetrahydrofolic acid, 5,10-
formiminotetrahydrofolic acid, 5-formyltetrahydrofolic acid (leucovorin), 10-
formyltetrahydrofolic acid, 10-methyltetrahydrofolic acid, one or more of the
folylpolyglutamates, compounds in which the pyrazine ring of the pterin moiety
of
folic acid or of the folylpolyglutamates is reduced to give dihydrofolates or

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-16-
tetrahydrofolates, or derivatives of all the preceding compounds in which the
N-5 or
N-10 positions carry one carbon units at various levels of oxidation, or
pharmaceutically compatible salts thereof, or a combination of two or more
thereof
Exemplary tetrahydrofolates include 5-formy1-(6S)-tetrahydrofolic acid, 5-
methyl-
(6S)-tetrahydrofolic acid, 5,10-methylene-(6R)-tetrahydrofolic acid, 5,10-
methenyl-
(6R)-tetrahydrofolic acid, 10-formy1-(6R)-tetrahydrofolic acid, 5-formimino-
(6S)-
tetrahydrofolic acid or (6S)-tetrahydrofolic acid, and pharmaceutically
acceptable
salts thereof Exemplary salts include sodium, potassium, calcium or ammonium
salts.
Exemplary relative weight ratios of BH4 to folates to arginine may be
from about 1:10:10 to about 10:1:1.
Optionally the stable formulations of the invention can also comprise
other excipients such as mannitol, hydroxyl propyl cellulose, microcrystalline

cellulose, of other non-reducing sugars such as sucrose, trehalose,
melezitose,
planteose, and raffinose. Reducing sugars may react with BH4.
Pharmaceutically acc;eptabit: ingredients for man uftiauring !roi;c1
formulations fcr oral administration may be incorporated, foJ: example, by -
rci!,)ixing
= the components and optionally finely dividing them, and then filling
capsules,
composed for example from hard or soft gelatin, compressing tablets, pills or
troches.
Coatings may be applied after compression to form pills.
Pharmaceutically acceptable ingredients are well known for the various
types of formulation and may be for example binders such as natural or
synthetic
polymers, excipients, lubricants, surfactants, sweetening and flavouring
agents,
coating materials, preservatives, dyes, thickeners, adjuvants, antimicrobial
agents,
antioxidants and carriers for the various formulation types. Nonlimiting
examples of
binders useful in a composition described herein include gum tragacanth,
acacia,
starch, gelatine, and biological degradable polymers such as homo- or co-
polyesters
of dicarboxylic acids, alkylene glycols, polyalkylene glycols and/or aliphatic
hydroxyl
carboxylic acids; homo- or co-polyamides of dicarboxylic acids, alkylene
diamines,
and/or aliphatic amino carboxylic acids; corresponding polyester-polyamide-co-
polymers, polyanhydrides, polyorthoesters, polyphosphazene and polycarbonates.
The

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-17-
biological degradable polymers may be linear, branched or crosslinked.
Specific
examples are poly-glycolic acid, poly-lactic acid, and poly-d,l-
lactide/glycolide. Other
examples for polymers are water-soluble polymers such as polyoxaalkylenes
(polyoxaethylene, polyoxapropylene and mixed polymers thereof, poly-
acrylamides
and hydroxylalkylated polyacrylamides, poly-maleic acid and esters or -amides
thereof, poly-acrylic acid and esters or -amides thereof, poly-vinylalcohol
und esters
or -ethers thereof, poly-vinylimidazole, poly-vinyl pyrrolidon, und natural
polymers
like chitosan.
Nonlimiting examples of excipients useful in a composition described
herein include phosphates such as dicalcium phosphate. Nonlimiting examples of
lubricants use in a composition described herein include natural or synthetic
oils, fats,
waxes, or fatty acid salts such as magnesium stearate.
Surfactants for use in a composition described herein can be anionic,
anionic, amphoteric or neutral. Nonlimiting examples of surfactants useful in
a
composition described herein include lecithin, phospholipids, octyl sulfate,
decyl
sulfate, dodecyl sulfate, tetradec:) sulfate, hexadecyl sulfate and. octadecyl
sulfate, Na
oleate or Na caprate, 1-acylantinccthalic-2-sulfonic acids, such as 1-
octanoylaminoethane-2-sulfonic acid. 1-decanoylaminoethane-2-sulfonic acid, 1-
dodecanoylaminoethane-2-sulfonic acid, 1-tetradecanoylaminoethane-2-sulfonic
acid,
1-hexadecanoylaminoethane-2-sulfonic acid, and 1-octadecanoylaminoethane-2-
sulfonic acid, and taurocholic acid and taurodeoxycholic acid, bile acids and
their
salts, such as choliC acid, deoxycholic acid and sodium glycocholates, sodium
caprate
or sodium laurate, sodium oleate, sodium lauryl sulphate, sodium cetyl
sulphate,
sulfated castor oil and sodium dioctyisulfosuccinate, cocamidopropylbetaine
and
laurylbetaine, fatty alcohols, cholesterols, glycerol mono- or -distearate,
glycerol
mono- or -dioleate and glycerol mono- or -dipalmitate, and polyoxyethylene
stearate.
Nonlimiting examples of sweetening agents useful in a composition
described herein include sucrose, fructose, lactose or aspartame. Nonlimiting
examples of flavoring agents for use in a composition described herein include
peppermint, oil of wintergreen or fruit flavors such as cherry or orange
flavor.
Nonlimiting examples of coating materials for use in a composition described
herein
include gelatin, wax, shellac, sugar or other biological degradable polymers.

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-18-
Nonlimiting examples of preservatives for use in a composition described
herein
include methyl or propylparabens, sorbic acid, chlorobutanol, phenol and
thimerosal.
The polymorph described herein may also be formulated as
effervescent tablet or powder, which disintegrate in an aqueous environment to
provide a drinking solution. Slow release formulations may also be prepared
from the
polymorph described herein in order to achieve a controlled release of the
active agent
=
in contact with the body fluids in the gastro intestinal tract, and to provide
a
substantial constant and effective level of the active agent in the blood
plasma. The
crystal form may be embedded for this purpose in a polymer matrix of a
biological
degradable polymer, a water-soluble polymer or a mixture of both, and
optionally
suitable surfactants. Embedding can mean in this context the incorporation of
micro-
particles in a matrix of polymers. Controlled release formulations are also
obtained
through encapsulation of dispersed micro-particles or emulsified micro-
droplets via
known dispersion or emulsion coating technologies.
The BH4 used in a composition described herein.is preferably
formulated as a dihydrochlorid..., sal t, iz is contemplated that other
salt forms
of BIM possess the desired biological ac-tivity, and consequently, ethersalt
forms of
BH4 can be used. Specifically, B114 salts with inorganic or organic acids are
preferred. Nonlimiting examples of alternative BH4 salts forms includes BH4
salts of
acetic acid, citric acid, oxalic acid, tartaric acid, fumaric acid, and
mandelic acid.
Pharmaceutically acceptable base addition salts may be formed with
metals or amines, such as alkali and alkaline earth metals or organic amines.
Pharmaceutically acceptable salts of compounds may also be prepared with a
pharmaceutically acceptable cation. Suitable pharmaceutically acceptable
cations are
well known to those skilled in the art and include alkaline, alkaline earth,
ammonium
and quaternary ammonium cations. Carbonates or hydrogen carbonates are also
possible. Examples of metals used as cations are sodium, potassium, magnesium,

ammonium, calcium, or ferric, and the like. Examples of suitable amines
include
isopropylamine, trimethylamine, histidine, N,N' dibenzylethylenediamine,
chloroprocaine, choline, diethanolamine, dicyclohexylamine, ethylenediamine, N
methylglucamine, and procaine.

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-19-
Pharmaceutically acceptable acid addition salts include inorganic or
organic acid salts. Examples of suitable acid salts include the
hydrochlorides,
acetates, citrates, salicylates, nitrates, phosphates. Other suitable
pharmaceutically
acceptable salts are well known to those skilled in the art and include, for
example,
acetic, citric, oxalic, tartaric, or mandelic acids, hydrochloric acid,
hydrobromic acid,
sulfuric acid or phosphoric acid; with organic carboxylic, sulfonic, sulfo or
phospho
acids or N substituted sulfarnic acids, for example acetic acid, propionic
acid, glycolic
acid, succinic acid, maleic acid, hydroxymaleic acid; methylmaleic acid,
fumaric acid,
malic acid, tartaric acid, lactic acid, oxalic acid, gluconic acid, glucaric
acid,
glucuronic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid,
salicylic acid,
4 aminosalicylic acid, 2 phenoxybenzoic acid, 2 acetoxybenzoic acid, embonic
acid,
nicotinic acid or isonicotinic acid; and with amino acids, such as the 20
alpha amino
acids involved in the synthesis of proteins in nature, for example glutamic
acid or
aspartic acid, and also with phenylacetic acid, methanesulfonic acid,
ethanesulfonic
acid, 2 hydroxyethanesulfonic acid, ethane 1,2 disulfCnic acid,
benzenesulfonic acid,
4 methylbenzenesulfoc acid, naphthalene 2, sulfonic acid, naphthalene 1,5
disulfonic
acid, 2 or 3 phosoliNlycerate, glucose.6 phosphate, N e,yclohexylsulfaniic
acid (With
the formation of cyclamates), or with other acid organic compounds, such as
ascorbic
acid.
The stable formulations of the invention may be provided, e.g. as
tablets or pills or capsules in HDPE bottles provided with a dessicant capsule
or
pouch; or in foil-on-foil blister packaging, or in blister packaging
comprising see-
through polymer film, if commercially desirable.
The phrase "pharmaceutically or pharmacologically acceptable" refers
to molecular entities and compositions that do not produce adverse; allergic,
or other
untoward reactions when administered to an animal or a human. As used herein,
"pharmaceutically acceptable carrier" includes any and all solvents,
dispersion media,
coatings, antibacterial and antifungal agents, isotonic and absorption
delaying agents
and the like. The use of such media and agents for pharmaceutically active
substances is well known in the art. Except insofar as any conventional media
or
agent is incompatible with the therapeutic compositions, its use in
therapeutic

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-20-
compositions is contemplated. Supplementary active ingredients also can be
incorporated into the compositions.
As used herein, "pharmaceutically acceptable carrier" includes any and
all solvents, dispersion media, coatings, antibacterial and antifungal agents,
isotonic
and absorption delaying agents and the like. The use of such media and agents
for
pharmaceutical active substances is well known in the art. Except insofar as
any
conventional media or agent is incompatible with the active ingredient, its
use in the.
therapeutic compositions is contemplated. Supplementary active ingredients
also can
be incorporated into the compositions.
It will be appreciated that the pharmaceutical compositions and .
treatment methods of invention may be useful in fields of human medicine and
veterinary medicine. Thus the subject to be treated may be a mammal,
preferably
human or other animal. For veterinary purposes, subjects include for example,
farm.
- animals including cowsõ sheep, pigs, horses and goats, companion animals,
such as
dogs and ea..s, exotic and/or zoo-animals,.laboratory animals
including.micerat
rabbits, L1Iiic. pigs and hamsters; and poultry such as chickens, trey
d;.1.c.ks and..
geese.
IN-. I've:,..17roarat Wthods Using Stable Formulations
The stable formulations of the invention may be Used for treatment of
conditions associated with elevated phenylalanine levels or decreased tyrosine
.or
tryptophan levels, which may be caused, for example, by reduced phenylalanine
hydroxylase, tyrosine hydroxylase, or tryptophan hydroxylase activity.
Conditions
associated with elevated phenylalanine levels specifically include
phenylketonuria,
both mild and classic, and hyperphenylalaninemia as described herein, and
exemplary
patient populations include the patient Subgroups described herein as well as
any other
.patient exhibiting phenylalanine levels above normal. Conditions associated
with
decreased tyrosine or tryptophan levels include neurotransmitter deficiency,
neurological and psychiatric disorders such as Parkinson's, dystonia,
spinocerebellar
degeneration, pain, fatigue, depression, other affective disorders and
schizophrenia.
The stable formulations may also be used for treating patients suffering
from BH4 deficiency, e.g., due to a defect in the pathway for its synthesis,
including

CA 02581814 2010-11-10
-21-
but not limited to dopa-responsive dystonia (DRD), sepiapterin reductase (SR)
deficiency, or dihydropteridine reductase (DHPR) deficiency.
Suitable subjects for treatment with the stable formulations of the
invention include subjects with an elevated plasma Phe concentration in the
absence
of the therapeutic, e.g. greater than 1800 M/L, or greater than 1600 p.M,
greater than
1400 M, greater than 1200 M, greater than 1000 M, greater than 800 1.tM, or

greater than 600 M, greater than 420 M, greater than 300 M, greater than
200 WA,
or greater than 180 M. Mild PKU is generally classified as plasma Phe
concentrations of up to 600pM/L, moderate PKU as plasma Phe concentrations .of
between 600 M/L to about 1200pM/L and classic or severe PKU as plasma Phe
concentrations that are greater than 12040\4/L. Preferably treatment with the
stable
formulations alone or with protein-restricted diet decreases the plasma
phenylalanine
concentration of the subject to less than 600 M, of less than 500 M, or 360
M
M or less, or less than 200 M, or less than 100 M. Other suitable subjects
15 include subjects diagnosed as having a reduced phenylalanine hydroxylase
(PAH)
activity. Reduced PAH activity may result from a mutation in the PAH enzyme,
for
example, a mutation in the catalytic domain of PAH or one or more mutations
selected from the group consisting of F39L, L48S, 165T, R68S, A104D, S1 10C,
D129G,E178G, V190A, P21 IT, R241C, R261Q, A300S, L308F, A3131, K.320N,
A373T, V388M E390G, A395P, P407S, and Y414C; or subjects that are pregnant.
females, females of child-bearing age that are contemplating pregnancy, or
infants
between 0 and 3 years of age, or 0-2, 0-1.5 or 0-1; or subjects diagnosed as
unresponsive within 24 hours to a single-dose BH4 loading test or a multiple
dose
loading test, such as a 4-dose or 7-day loading test. Exemplary patient
populations
and exemplary 8H4 loading tests are described in Intl. Publication No. WO
2005/049000.
U.S. Patent Nos. 4,752,573; 4,758,571; 4,774,244; 4,920,122;
5,753,656; 5,922,713; 5,874,433; 5,945,452; 6,274,581; 6,410,535; 6,441,038;
6,544,994; and U.S. Patent Publications US 20020187958; US 20020106645; US
2002/0076782; US 20030032616 each describe methods of administering BH4
compositions for non-PKU treatments. Each of those patents provides a general
teaching of

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-22-
methods of administering BH4 compositions known to those of skill in the art,
that
may be adapted for the treatment as described herein.
While individual needs vary, determination of optimal ranges of
effective amounts of each component is within the skill of the art. Typical
dosages of
the B114 comprise about 1 to about 20 mg/kg body weight per day, which will
usually
amount to about 5 (1 mg/kg x 5kg body weight) to 3000 mg/day (30mg/kg x 100kg
body weight). While continuous, daily administration is contemplated, for HPA
it
may be desirable to cease the BH4 therapy when the symptoms of Phe levels are
reduced to below a.certain threshold level. Of course, the therapy may be
reinitiated
in the event that Phe levels rise again. Appropriate dosages may be
ascertained
through the use of established assays for determining blood levels of Phe in
conjunction with relevant dose response data.
In preferred embodiments, it is contemplated that the methods of the
:present invention will provide to a patient in need thereof, a daily dose of
between
about 10 mg/kg to about 20 mg/kg of BH4. Of course, one skilled,in thenrt may.
adjust. this dose up or dpwn depending on the efficacy being achieved-by the
administration. The daily dose may ;3e administered ',11 a sirtgle dose. or
alternative4,
may be administered in multiple doses at conveniently spaced intervals. In
exemplary
embodiments, the daily dose may be 1 mg/kg; 2 mg/kg, 3 mg/kg, 4 mg/kg,
5.mg/kg, 6
mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 1.4
mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg, 20 mg/kg, 22 mg/kg,
24
mg/kg, 26 mg/kg, 28 mg/kg, 30 mg/kg, 32 mg/kg, 34 mg/kg, 36 mg/kg, 38 mg/kg,
40
mg/kg, 42 mg/kg, 44 mg/kg, 46 mg/kg, 48 mg/kg, 50 mg/kg, or more mg/kg.
The invention further contemplates that stable formulations of the
invention may be used for treatment of subjects suffering from conditions that
would
benefit from enhancement of nitric oxide synthase activity and patients
suffering from
vascular diseases, ischemic or inflammatory diseases, or insulin resistance.
The
treatment may, for example alleviate a deficiency in nitric oxide synthase
activity or
may, for example provide an increase in nitric oxide synthase activity over
normal
levels. It has been suggested that a patient suffering from a deficiency in
nitric oxide
synthase activity would benefit from co-treatment with folates, including
folate
precursors, folic acids, or folate derivatives.

CA 02581814 2010-11-10
-23-
Nitric oxide is constitutively produced by vascular endothelial cells
where it plays a key physiological role in the regulation of blood pressure
and
vascular tone. It has been suggested that a deficiency in nitric oxide
bioactivity is
involved in the pathogenesis of vascular dysfunctions, including coronary
artery
disease, atherosclerosis of any arteries, including coronary, carotid,
cerebral, or
peripheral vascular arteries, ischemia-reperfusion injury, hypertension,
diabetes,
diabetic vasculopathy, cardiovascular disease, peripheral vascular disease, or

neurodegenerative conditions stemming from ischemia and/or inflammation, such
as
stroke, and that such pathogenesis includes damaged endothelium, insufficient
oxygen
flow to organs and tissues, elevated systemic vascular resistance (high blood
pressure), vascular smooth muscle proliferation, progression of vascular
stenosis
(narrowing) and inflammation. Thus, treatment of any of these conditions is
contemplated according to methods of the invention.
It has also been suggested that the enhancement of nitric oxide
synthase activity also results in reduction of elevated superoxide levels,
increased
insulin sensitivity, and reduction in vascular dysfunction associated with
insulin resistance, as described in U.S. Patent No. 6,410,535. Thus,
treatment of diabetes (type 1 or type II), hyperinsulinemia, or insulin
resistance is contemplated according to the invention. Diseases having
vascular
dysfunction associated with insulin resistance include those caused by insulin
resistance or aggravated by insulin resistance, or those for which cure is
retarded by
insulin resistance, such as hypertension, hyperlipidemia, arteriosclerosis,
coronary
vasoconstrictive angina, effort angina, cerebrovascular constrictive lesion,
cerebrovascular insufficiency, cerebral vasospasm, peripheral circulation
disorder,
coronary arteriorestenosis following percutaneous transluminal coronary
angioplasty
(PTCA) or coronary artery bypass grafting (CAB 0), obesity, insulin-
independent
diabetes, hyperinsulinemia, lipid metabolism abnormality, coronary
arteriosclerotic
heart diseases or the like so far as they are associated with insulin
resistance. It is
contemplated that when administered to patients with these diseases, BH4 can
prevent
or treat these diseases by activating the functions of NOS, increasing NO
production
and suppressing the production of active oxygen species to improve disorders
of
vascular endothelial cells.

CA 02581814 2010-11-10
-24-
It is understood that the suitable dose of a composition according to the
present invention will depend upon the age, health and weight of the
recipient, kind of
concurrent treatment, if any, frequency of treatment, and the nature of the
effect
desired (L e., the amount of decrease in plasma Phe concentration desired).
The
frequency of dosing also is dependent on pharmacodynamic effects on Phe
levels. If
the effect lasts for 24 hours from a single dose. However, the most preferred
dosage
can be tailored to the individual subject, as is understood and determinable
by one of
skill in the art, without undue experimentation. This typically involves
adjustment of
a standard dose, e.g., reduction of the dose if the patient. has a low body
weight.
The frequency of BH4 dosing will depend on the pharmacokinetic
parameters of the agent and the routes of administration. The optimal
pharmaceutical
formnlation will be determined,by one of skill in the art depending on the
route of
administration and the desired dosage. See fix example Remington's
Pharmaceutical
Sciences, 18th Ed. (1990, Mack Publ. Co, Easton PA 18042) pp 1435 1712.
Such formulations may influence the physical state, stability, rate of in vivo
release and rate of in vivo clearance of the administered agents. Depending on
the route of administration, a suitable dose may be calculated according to
body weight, body surface areas or organ size. Further refinement of the
calculations necessary to determine the appropriate treatment dose is
routinely made
by those of ordinary skill in the art without undue experimentation,
especially in light
of the dosage information and assays disclosed herein as well as the
phaimacokinetic
data observed in animals or human clinical trials.
The final dosage regimen will be determined by the attending
physician, considering factors which modify the action of drugs, e.g., the
drug's
specific activity, severity of the damage and the responsiveness of the
patient, the age,
condition, body weight, sex and diet of the patient, the severity of any
infection, time
of administration and other clinical factors. As studies are conducted,
further
information will emerge regarding appropriate dosage levels and duration of
treatment
for specific diseases and conditions.
V. Combination therapy

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-25-
Certain methods of the invention involve the combined use of the
stable formulations of the invention and one or more other therapeutic agents.
In such combination therapy, administration of the stable formulations
of the invention may be concurrent with or may precede or follow the
administration
of the second therapeutic agent, e.g. by intervals ranging from minutes to
hours, so
long as both agents are able to exert their therapeutic effect at overlapping
time
periods. Thus, the invention contemplates the stable formulations of the
invention for
use with a second therapeutic agent. The invention also contemplates use of a
second
therapeutic agent in preparation of a medicament for administration with the
stable
tetrahydrobiopterin, precursor, derivative or analog formulations of the
invention.
Tetrahydrobiopterin therapy may be combined with dietary protein
restriction to effect a therapeutic outcome in patients with various forms of
HPA. For
example, one could administer to the subject the BH4 composition and a low-
phenylalanine medical protein composition in a combined amount effective to
produce the desired therapeutic outcome (i.e., .a lowering of plasma Phe
concentration,
andkr the ability to tolerate greater atroinits of Phe/protein intake without
producing
a concomitant increase in plasma Phe concentrations). This proceS:s ..,nay
involve
administering the BH4 composition and the dietary prpteia therapeutic
composition at:
the same time. This may be achieved by administering a single composition or
pharmacological protein formulation that includes all of the dietary protein
requirements and also includes the BIM within said protein formulation.
Alternatively, the dietary protein (supplement or normal protein meal) is
taken at
about the same time as a pharmacological formulation (tablet, injection or
drink) of
BH4.
In other alternatives, the BH4 treatment may precede or follow the
dietary protein therapy by intervals ranging from minutes to hours. In
embodiments
where the protein and the BH4 compositions are administered separately, one
would
generally ensure that a significant period of time did not expire between the
time of
each delivery, such that the BH4 will still be able to exert an advantageously
effect on
the patient. In such instances, it is contemplated that one would administer
the BH4
within about 2-6 hours (before or after) of the dietary protein intake, with a
delay time
of only about 1 hour being most preferred. In certain embodiments, it is
contemplated

CA 02581814 2007-03-22
WO 2006/055511
PCT/US2005/041252
-26-
that the BH4 therapy will be a continuous therapy where a daily dose of BH4 is

administered to the patient indefinitely. In other situations, e.g., in
pregnant women
having only the milder forms of PKU and HPA, it may be that the BH4 therapy is

only continued for as long as the woman is pregnant and/or breast feeding.
Further, in addition to therapies based solely on the delivery of BH4
and dietary protein regulation, the methods of the present invention also
contemplate
combination therapy with a third composition that specifically targets one or
more of
the symptoms of HPA. For example, it is known that the deficit in tyrosine
caused by
HPA results in a deficiency in neurotransmitters dopamine and serotonin. Thus,
in
the context of the present invention, it is contemplated that BH4 and dietary
protein
based methods could be further combined with administration of L-dopa,
carbidopa
and 5-hydroxytryptophan neurotransmitters to correct the defects that result
from
decreased amounts of tyrosine in the diet.
In addition, gene therapy with both PAH (Christensen et al., Mol.
Gent. And Metabol. 76: 313-318, 2002; Christensen et al., Gene Therapy,
7:1971.
1978, 2000) and ph,:-:nylalanine ammonia-lyase (PAL Liu et al:,-Arts: Cells.
Blood.
Subs and Irnmob. Bi wch. 30(4)243-257, 2002) has been contemplated by the of
skill in the art. Such gene therapy techniques could be used in combination
with the
combined BH4/dietary protein restriction based therapies of the invention. In
further
combination therapies, it iS contemplated that phenylase may be provided as an
injectable enzyme to destroy lower Phe concentrations in the patient. As the
administration of phenylase would not generate tyrosine (unlike administration
of
PAH), such treatment will still result in tyrosine being an essential amino
acid for
such patients. Therefore dietary supplementation with tyrosine may be
desirable for
patients receiving phenylase in combination with the BH4 therapy.
VII. Examples
The following examples are included to demonstrate preferred
embodiments of the invention. It should be appreciated by those of skill in
the art that
the techniques disclosed in the examples which follow represent techniques
discovered by the inventor to function well in the practice of the invention,
and thus

CA 02581814 2013-09-10
-27-
can be considered to constitute preferred modes for its practice.
EXAMPLE 1
Preparation of Stabilized Crystallized Form of 13II4
Int'l. Publication No. WO 2005/065018 describes X ray and Raman
spectra studies to characterize the polymorphs, including hydrates or
solvates, of BH4,
as well as exemplary crystallization conditions under which the polymorphs can
be
prepared. Int'l. Publication No. WO 2005/049000 describes various patient
populations
for which BH4 treatment is suitable and describes methods for treating such
subjects
with BH4. Int'l. Publication No. WO 2005/049614 describes methods of
synthesizing
BH4.
20 EXAMPLE 2
Stable Tablet Formulation of Tetrahydrobiopterin
Three tablet formulations were prepared by mixing the ingredients
shown in Table 1 as described in detail below.
Table I
Formulation I Formulation II Formulation III
Ingredient (wt %) (wt %) (wt %)
6R-L-etythro-5, 6, 7, 8-tetrahydrobiopterin
dihydrochloride salt, polymozph B 33.33 33.33 33.33
(Active Ingredient)
D-Mannitol
57.56 57.56 57.56
(Taste Masking)

CA 02581814 2010-11-10
-28--
Dibasic Calcium Phosphate, Anhydrous
2.18 2.18 2.18
(Binder)
Hydroxypropyl Cellulose
3.63 4.5
(Disintegrant)
Polyvinylpyrrolidone
0.87 4.50
(Disintegrant)
Ascorbic acid
1.67 1.67 1.67
(Stabilizer)
Riboflavin
0.01 0.01 0.01
(Coloring Agent)
Sodium Stearyl Fumarate
0.75 0.75 0.75
(Lubricant)
For each formulation in Table I, twelve kilogram batches were
prepared by first charging 4 kg of 6R-L-erythro-5, 6, 7, 8-tetrahydrobiopterin

dihydrochloride salt (SAPROPTER1N1m Hydrochloride, available from Daiichi
Suntory
Phstim.: Cc., Ltd., Japan to a blender and blending the BH4 for 10 minutes at
25
revolutions per minute (RPM). Then 6.91 kg of D-Mannitol (PEARLITOLTm,
available
from Roquette America, nc., Keokuk. Iowa) was added to the blender and thc
mixture was allowed to blend for an additional 10 minutes at 25 RPM. Then 260
grams of Anhydrous Dibasic Calcium Phosphate (available from Mallinckrodt
Baker,
Inc., Phillipsburg, New Jersey) and (a) in Formulation I, 436 grams of
Hydroxypropyl
Cellulose and 104 grams were added to the blender, (b) in Formulation II 540
grams
of Hydroxypropyl Cellulose was added to the blender; (c) in Formulation III,
540
grams of Polyvinylpyrrolidone (KOLLIDONTM CL, available from BASF Corporation,

Florham Park, New Jersey) were added to the blender, and the mixture was
allowed to
blend for an additional 10 minutes at 25 RPM. To the bender 200 grams of
Ascorbic
Acid and 120 grams of Ribofloavin were added to the blender and the mixture
was
allowed to blend for 3 minutes at 25 RPM. The Sodium Stearyl Fumarate
lubricant
(PRUVTm, available from Penwest Pharmaceuticals Co., Danbury, Connecticut) was

filtered through a 25 mesh stainless steel screen and into a bag, and the
blender was
then charged with 9 kg of the screened Sodium Stearyl Fumarate, and the
resulting
mixture was allowed to blend for 5 minutes at 25 RPM.

CA 02581814 2007-03-22
WO 2006/055511
PCT/US2005/041252
-29-
The blended mixture of each formulation were then removed from the
blender, and three samples of each formulation were collected for the
preparation of a
150 mg, a 300 mg, and a 600 mg tablets. For each formulation, the tablet
samples
(150 mg, 300 mg, and 600 mg) were placed in a tablet press (available from
Jenn-
Chiang Mahinery Co., Ltd., Taiwan, R.O.C.) wherein the parameters of the
tablet
press were set to provide tablets with a thickness in the range of 4.5 to 5.5
millimeters, and a target hardness of 7 KP.
The resulting tablets were then analyzed to determine the stability of
the formulations. The stability of the formulations was studied for a change
in
appearance over time by a visual inspection at different intervals, for
disintegration of
the formulation utilizing the United States Pharmacopeia recommendations no.
701,
and for a chemical change by assaying the components of the formulations.
The.),
results of the stability tests are summarized below in Table II.
Table 11
Form. ft Initiai - 2. weeks 4'weeks 8
wet)cs
= Dark
Dark yellow Dark yellow
yellow
spots spots
Off white,spots
(diameter 1- (diameter 1-
Appearance round
(diameter
2mm) on the 2mm) on
Tablets1-2mm) on
off white the off white
the off
tablets tablets
white tablet
5 min 20
Disintegration 5 min 40 sec 8 min 4 sec
sec
Chemical
97.10% 97.90% 98.2 100.7
Assay
II Off white
Broken chip Broken chip Broken
round on some on some chip
on
Tablets tablets. tablets. some
Light yellow Light yellow tablets.
Appearance spots spots Light
(diameter 1- (diameter 1- brown
= 2 mm) on 2 mm) on spots
the off white the off white (diameter
round round 1-2 mm) on

CA 02581814 2007-03-22
WO 2006/055511
PCT/US2005/041252
-30-
tablets. tablets the
dark
brown
tablets
4 mm 10 4 min 38 sec 6 min 52 sec
Disintegration
sec
Chemical 102.70% 100.00% 100.2 97.4
Assay
= Rough Rough
Rough
Color is surface, and surface, and
surface,
Appearance
off white color is light color is light and color
is
yellow yellow yellow
1 min 52
Disintegration 35 sec 58 sec
sec
Chemical
100.20% 102.90% 97.4 =
99.8
Assay
The stability tests show that tablet Formulation lU is more stablediat
the other formulations of BH4. Each of the pharmaceutical preparations are
usefgt
formlations for the delivery of BH4. Formulation III exhibiLcd better
stability. t1i:4n
Fdrinpia tions I and II. Thus, in one preferred embodiment, thctabizedtabiet
formulation comprises an optimal disintegration agent, for example,
crossprovidone
.or .a disintegration. agent more similar to polyvinylpyrrolidone than
hydroxypropylcellulose. The preferred formulation is Formulation III. Other
suitable
tablet formulations may include at least ascorbic acid at a concentration of
at least
0.01% weight, or at least 0.05% weight or at least 0.1% weight.
EXAMPLE 3
Three hundred mg tablets containing 100 mg tetrahydrobiopterin are
prepared using the desired initial amount of polymorph B and mixing with other

ingredients in the relative amounts shown below in Table III using the
following dry
tableting process. Tablets containing other desired amounts of
tetrhydrobiopterin can
be prepared in a similar manner.
The 6R-L-erythro-5, 6, 7, 8-tetrahydrobiopterin and D-Mannitol were
hand screened with a 20 mesh screen (designed to filter out particles greater
than
--850 pm in size) and placed in a blender. The mixture was blended for 10
minutes at

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-31-
21 RPM. Next, the Anhydrous Dibasic Calcium Phosphate and CROSPOVIDONE
were hand screened with a 20 mesh screen and blended with the BH4 and D-
Mannitol
for 10 minutes at 21 RPM. The Ascorbic Acid and Riboflavin were added to the
blender after hand screening with a 20 mesh screen, and the resulting mixture
blended
for 10 minutes at 21 RPM. Next, the Sodium Stearyl Fumarate was added to the
blender after hand screening with a 40 mesh screen, and blended for 5 minutes
at 21
RPM. The blended mixture was then discharged into a bag and tested for
uniformity,
before being pressed into tablets.
Table III
(wt %) (mg)
Ingredient
6R-L-erythro-5, 6, 7, 8-tetrahydrobiopterin
dihydrochloride salt 33.33 100.0
(Active Ingredient)
D-Mannitol
57.56 172.68
(Taste Masking)
Dibasic Calcium Phosphate, Anhydrous
2.18 6.54
(Binder)
CROSPOVIDONE
4.50 13.50
(Disintegration Agent)
Ascorbic acid
1.67 5.00
(Acidic Antioxidant)
Riboflavin
0.01 0.03
(Coloring Agent)
Stearyl Fumarate
0.75 2.25
(Lubricant)
The tablets were packaged either in foil blister-packs, or in HDPE
bottles in quantities of 45-tablets per bottle. Each type of packaged tablets
were
divided into two batches. One batch was stored at room temperature, 25+2 C,
and
60+5% relative humidity. The other batch was stored under accelerated testing
conditions, at 40+2 C and 75+5% relative humidity. At regular intervals,
tablets were
removed from storage and tested for retention of the active pharmaceutical
ingredient,

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-32-
6R-L-erythro-5, 6, 7, 8-tetrahydrobiopterin. Exemplary results for the 300 mg
tablets
are shown below in Tables IV, V, VI and VII. After six months storage under
room
temperature or accelerated testing conditions, each of the four batches showed

retention of at least 99% of the original amount of 6R-L-erythro-5, 6, 7, 8-
tetrahydrobiopterin by HPLC assay, loss on drying of less than 1%, and rapid
disintegration in 3 minutes or less.
Table IV
SAPROPTE1RIN DIHYDROCHLORIDE DRUG PRODUCT
(300-MG TABLET, FOIL BLISTER-PACK)
STORAGE CONDITIONS: 25 2 C / 60 5%RH
Time Point (months)
Test/Assay Stability Specification 1 3 6 9
Appearance
White to light yellow
by Visual Conforms Conforms Conforms Conforms Conforms
compressed tablets
Inspection
Loss on
Report Result 0.3% 0.3% 0.9% 0.7% 0.9%
Drying
1 min. 2 min. 2 min. 2 min. , 1
min.
Disintegration 3 minutes
30 sec. 7 sec. 2 sec. 35 sec. 48
sec.
HPLC Assay 90 - 110% 101% 100% 100% 100% 101%
0.03%
(RRT. 0.69)
0.01% =
0.05% 0.09% -(RRT 0.73)
(RRT 0.72) (RRT 0.81) 0.07%
individual 0.04% 0.08% 0.04% (RRT
0.81)
0.05%
unidentified (RRT 1.28) (RRT 0.83) (RRT
0.83) = .:, 0.03%
HPLC for 0.05% 0.06% (ART
0.83)
Related (RRT 1.32) (RRT
1.24) 0.02%
Substances (RRT 1.21)
0.04%
(RRT 1.44)
5 0.5% total
0.05% 0.04% 0.18% 0.22% 0.20%
unidentified
2.0 % total related 0.05% 0.04% 0.18% 0.22% 0.20%
substances
ND = None detected

CA 02581814 2007-03-22
WO 2006/055511 PCT/US2005/041252
-33-
Table V
SAPROPTERIN DIHYDROCHLORIDE DRUG PRODUCT
(300-MG TABLET, FOIL BLISTER-PACK)
STORAGE CONDITIONS: 40 2 C / 75 5 /oRH
Time Point (months)
Test/Assay Stability Specification
0 1 3 6
Appearance by Visual White to light yellow compressed
Conforms Conforms Conforms Conforms
Inspection tablets
Loss on Drying Report Result 0.3% 0.3% 0.8%
0.7%
1 min. 2 min. 2 min. 2
min.
Disintegration 3 minutes
30 sec. 16 sec. 47 sec.
11 sec.
HPLC Assay 90 ¨ 110% 101% 100% 101%
99%
0.06% 0.07%
(RRT 0.64) (RRT 0.74)
0.07% 0.15%
0.04%
< 0.1% individual unidentified 0.05% (RRT 1.28)
(RRT 0.72) (RRT 0.83)
HPLC for Related 0.04%
0.04%
Substances (RRT 0.83)
(RRT 0.83)
0.06% 0.06%
(RRT 1.31) (RRT 1.24)
< 0.5% total unidentified 0.05% 0.04% 0.23%
0.32%
< 2.0 % total related substances 0.05% 0.04% 0.23%
0.32%
ND = None detected

CA 02581814 2007-03-22
WO 2006/055511
PCT/US2005/041252
-34-
Table VI
SAPROPTERIN DIHYDROCHLORIDE DRUG PRODUCT
(300-MG TABLET, 45 TABLET BOTTLES)
STORAGE CONDITIONS: 25 2 C / 60 5%R1I
Time point (months)
Test/Assay Stability Specification
0 3 6
Appearance
White to light yellow
by Visual Conforms Conforms Conforms
compressed tablets
Inspection
Loss on
Report Result 0.7% 0.8% 1.1%
Drying
2 min 2 min 2 min
Disintegration 5 3 minutes
57 sec 3 sec 50 sec
HPLC Assay 90 ¨ 110% 102% NT 103%
0.08%
(RRT 0.56)
0.04%
5 0.1% individual (RRT 0.61)
0.06% NT
unidentified 0.05%
(RRT0.65)
0.04%
HPLC for (RRT 0.81)
Related 5 0.5% total
0.15% NT 0.21%
Substances unidentified
5 2.0 % total related
0.15% NT 0.21%
substances
Report Result (mg/tablet) 5 NT 5
Total Aerobic Microbial
< 100 cfu/g NR NR
Count 5. 1000 cfu/g
Test for Escherichia coli =
Absent
absent
ND =None detected
NT = Not tested
NR = Not required
=

CA 02581814 2007-03-22
WO 2006/055511
PCT/US2005/041252
-35-
Table VII
SAPROPTERIN DIHYDROCHLORIDE DRUG PRODUCT
(300-MG TABLET, 45-TABLET BOTTLES)
STORAGE CONDITIONS: 40 2 C / 75 5 VoRH
Time Point (months)
Test/Assay Stability Specification
0 3 6
Appearance by Visual White to light yellow compressed
Conforms Conforms Conforms
Inspection tablets
Loss on Drying Report Result 0.7% 1.1% 1.1%
2 min 2 min 2 min
Disintegration < 3 minutes
57 sec 41 sec 48 sec
HPLC Assay 90¨ 110% 102% NT 101%
0.13%
(RRT 0.56)
0.03%
< 0.1% individual unidentified 0.06% NT (RRT
0.61)
0.10%
HPLC for Related (RRT
0.65)
Substances 0.04%
(RRT 0.81)
< 0.5% total unidentified 0.15% NT 0.30%
< 2.0% total related substances 0.15% NT 0.30%
Report Result (mg/tablet) 5 NT 5
ND = None detected
NT = Not tested
NR = Not required

Representative Drawing

Sorry, the representative drawing for patent document number 2581814 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2015-10-13
(86) PCT Filing Date 2005-11-16
(87) PCT Publication Date 2006-05-26
(85) National Entry 2007-03-22
Examination Requested 2010-11-10
(45) Issued 2015-10-13

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $473.65 was received on 2023-10-19


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if standard fee 2024-11-18 $624.00
Next Payment if small entity fee 2024-11-18 $253.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Registration of a document - section 124 $100.00 2007-03-22
Application Fee $400.00 2007-03-22
Maintenance Fee - Application - New Act 2 2007-11-16 $100.00 2007-11-05
Maintenance Fee - Application - New Act 3 2008-11-17 $100.00 2008-11-03
Maintenance Fee - Application - New Act 4 2009-11-16 $100.00 2009-11-04
Maintenance Fee - Application - New Act 5 2010-11-16 $200.00 2010-11-04
Request for Examination $800.00 2010-11-10
Maintenance Fee - Application - New Act 6 2011-11-16 $200.00 2011-11-02
Maintenance Fee - Application - New Act 7 2012-11-16 $200.00 2012-10-30
Maintenance Fee - Application - New Act 8 2013-11-18 $200.00 2013-10-31
Maintenance Fee - Application - New Act 9 2014-11-17 $200.00 2014-11-13
Final Fee $300.00 2015-06-17
Maintenance Fee - Patent - New Act 10 2015-11-16 $250.00 2015-11-09
Maintenance Fee - Patent - New Act 11 2016-11-16 $250.00 2016-11-14
Maintenance Fee - Patent - New Act 12 2017-11-16 $250.00 2017-11-13
Maintenance Fee - Patent - New Act 13 2018-11-16 $250.00 2018-11-12
Maintenance Fee - Patent - New Act 14 2019-11-18 $250.00 2019-11-08
Maintenance Fee - Patent - New Act 15 2020-11-16 $450.00 2020-11-06
Maintenance Fee - Patent - New Act 16 2021-11-16 $459.00 2021-11-12
Maintenance Fee - Patent - New Act 17 2022-11-16 $458.08 2022-10-24
Maintenance Fee - Patent - New Act 18 2023-11-16 $473.65 2023-10-19
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
BIOMARIN PHARMACEUTICAL INC.
Past Owners on Record
BAFFI, ROBERT
HENDERSON, MARK
JUNGLES, STEVEN
SLUZKY, VICTORIA
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2007-03-22 1 55
Claims 2007-03-22 5 198
Drawings 2007-03-22 1 17
Description 2007-03-22 35 1,705
Cover Page 2007-05-24 1 26
Description 2010-11-10 35 1,685
Claims 2010-11-10 6 206
Description 2012-11-02 36 1,721
Claims 2012-11-02 7 236
Claims 2013-09-10 7 234
Description 2013-09-10 36 1,718
Claims 2014-07-30 7 243
Description 2014-07-30 36 1,728
Cover Page 2015-10-05 1 26
PCT 2007-03-22 3 81
Assignment 2007-03-22 4 103
Correspondence 2007-05-18 1 27
Prosecution-Amendment 2010-11-10 16 669
Assignment 2007-09-05 12 449
Prosecution-Amendment 2012-05-03 2 70
Prosecution-Amendment 2012-11-02 24 913
Prosecution-Amendment 2013-09-10 11 374
Prosecution-Amendment 2014-07-30 5 184
Prosecution-Amendment 2013-03-12 2 54
Prosecution-Amendment 2014-01-31 2 49
Final Fee 2015-06-17 1 39